Skip to main content
Top
Published in: Diabetologia 3/2015

01-03-2015 | Review

Epigenetic mechanisms in diabetic complications and metabolic memory

Authors: Marpadga A. Reddy, Erli Zhang, Rama Natarajan

Published in: Diabetologia | Issue 3/2015

Login to get access

Abstract

The incidence of diabetes and its associated micro- and macrovascular complications is greatly increasing worldwide. The most prevalent vascular complications of both type 1 and type 2 diabetes include nephropathy, retinopathy, neuropathy and cardiovascular diseases. Evidence suggests that both genetic and environmental factors are involved in these pathologies. Clinical trials have underscored the beneficial effects of intensive glycaemic control for preventing the progression of complications. Accumulating evidence suggests a key role for epigenetic mechanisms such as DNA methylation, histone post-translational modifications in chromatin, and non-coding RNAs in the complex interplay between genes and the environment. Factors associated with the pathology of diabetic complications, including hyperglycaemia, growth factors, oxidant stress and inflammatory factors can lead to dysregulation of these epigenetic mechanisms to alter the expression of pathological genes in target cells such as endothelial, vascular smooth muscle, retinal and cardiac cells, without changes in the underlying DNA sequence. Furthermore, long-term persistence of these alterations to the epigenome may be a key mechanism underlying the phenomenon of ‘metabolic memory’ and sustained vascular dysfunction despite attainment of glycaemic control. Current therapies for most diabetic complications have not been fully efficacious, and hence a study of epigenetic mechanisms that may be involved is clearly warranted as they can not only shed novel new insights into the pathology of diabetic complications, but also lead to the identification of much needed new drug targets. In this review, we highlight the emerging role of epigenetics and epigenomics in the vascular complications of diabetes and metabolic memory.
Literature
1.
go back to reference Beckman JA, Creager MA, Libby P (2002) Diabetes and atherosclerosis: epidemiology, pathophysiology, and management. JAMA 287:2570–2581PubMed Beckman JA, Creager MA, Libby P (2002) Diabetes and atherosclerosis: epidemiology, pathophysiology, and management. JAMA 287:2570–2581PubMed
2.
go back to reference Ziyadeh FN, Sharma K (2003) Overview: combating diabetic nephropathy. J Am Soc Nephrol 14:1355–1357PubMed Ziyadeh FN, Sharma K (2003) Overview: combating diabetic nephropathy. J Am Soc Nephrol 14:1355–1357PubMed
3.
go back to reference Fong DS, Aiello L, Gardner TW et al (2003) Diabetic retinopathy. Diabetes Care 26:226–229PubMed Fong DS, Aiello L, Gardner TW et al (2003) Diabetic retinopathy. Diabetes Care 26:226–229PubMed
4.
go back to reference Natarajan R, Nadler JL (2004) Lipid inflammatory mediators in diabetic vascular disease. Arterioscler Thromb Vasc Biol 24:1542–1548PubMed Natarajan R, Nadler JL (2004) Lipid inflammatory mediators in diabetic vascular disease. Arterioscler Thromb Vasc Biol 24:1542–1548PubMed
5.
go back to reference Vincent AM, Calabek B, Roberts L, Feldman EL (2013) Biology of diabetic neuropathy. Handb Clin Neurol 115:591–606PubMed Vincent AM, Calabek B, Roberts L, Feldman EL (2013) Biology of diabetic neuropathy. Handb Clin Neurol 115:591–606PubMed
6.
go back to reference Forbes JM, Cooper ME (2013) Mechanisms of diabetic complications. Physiol Rev 93:137–188PubMed Forbes JM, Cooper ME (2013) Mechanisms of diabetic complications. Physiol Rev 93:137–188PubMed
7.
go back to reference Brownlee M (2001) Biochemistry and molecular cell biology of diabetic complications. Nature 414:813–820PubMed Brownlee M (2001) Biochemistry and molecular cell biology of diabetic complications. Nature 414:813–820PubMed
8.
go back to reference Kato M, Natarajan R (2014) Diabetic nephropathy—emerging epigenetic mechanisms. Nat Rev Nephrol 10:517–530PubMed Kato M, Natarajan R (2014) Diabetic nephropathy—emerging epigenetic mechanisms. Nat Rev Nephrol 10:517–530PubMed
9.
go back to reference Woroniecka KI, Park AS, Mohtat D, Thomas DB, Pullman JM, Susztak K (2011) Transcriptome analysis of human diabetic kidney disease. Diabetes 60:2354–2369PubMedCentralPubMed Woroniecka KI, Park AS, Mohtat D, Thomas DB, Pullman JM, Susztak K (2011) Transcriptome analysis of human diabetic kidney disease. Diabetes 60:2354–2369PubMedCentralPubMed
10.
go back to reference Dunham I, Kundaje A, Aldred SF et al (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489:57–74 Dunham I, Kundaje A, Aldred SF et al (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489:57–74
11.
go back to reference Human Epigenome Task Force (2008) Moving AHEAD with an international human epigenome project. Nature 454:711–715 Human Epigenome Task Force (2008) Moving AHEAD with an international human epigenome project. Nature 454:711–715
12.
go back to reference Kanwar YS, Sun L, Xie P, Liu FY, Chen S (2011) A glimpse of various pathogenetic mechanisms of diabetic nephropathy. Annu Rev Pathol 6:395–423PubMedCentralPubMed Kanwar YS, Sun L, Xie P, Liu FY, Chen S (2011) A glimpse of various pathogenetic mechanisms of diabetic nephropathy. Annu Rev Pathol 6:395–423PubMedCentralPubMed
13.
go back to reference Geraldes P, King GL (2010) Activation of protein kinase C isoforms and its impact on diabetic complications. Circ Res 106:1319–1331PubMedCentralPubMed Geraldes P, King GL (2010) Activation of protein kinase C isoforms and its impact on diabetic complications. Circ Res 106:1319–1331PubMedCentralPubMed
14.
go back to reference Ramasamy R, Yan SF, Schmidt AM (2011) Receptor for AGE (RAGE): signaling mechanisms in the pathogenesis of diabetes and its complications. Ann N Y Acad Sci 1243:88–102PubMed Ramasamy R, Yan SF, Schmidt AM (2011) Receptor for AGE (RAGE): signaling mechanisms in the pathogenesis of diabetes and its complications. Ann N Y Acad Sci 1243:88–102PubMed
15.
16.
go back to reference Averill MM, Bornfeldt KE (2009) Lipids versus glucose in inflammation and the pathogenesis of macrovascular disease in diabetes. Curr Diabetes Rep 9:18–25 Averill MM, Bornfeldt KE (2009) Lipids versus glucose in inflammation and the pathogenesis of macrovascular disease in diabetes. Curr Diabetes Rep 9:18–25
17.
go back to reference Sanchez AP, Sharma K (2009) Transcription factors in the pathogenesis of diabetic nephropathy. Expert Rev Mol Med 11:e13PubMed Sanchez AP, Sharma K (2009) Transcription factors in the pathogenesis of diabetic nephropathy. Expert Rev Mol Med 11:e13PubMed
18.
go back to reference Villeneuve LM, Natarajan R (2010) The role of epigenetics in the pathology of diabetic complications. Am J Physiol Ren Physiol 299:F14–F25 Villeneuve LM, Natarajan R (2010) The role of epigenetics in the pathology of diabetic complications. Am J Physiol Ren Physiol 299:F14–F25
19.
go back to reference Writing Team for the DCCT/EDIC Research Group (2002) Effect of intensive therapy on the microvascular complications of type 1 diabetes mellitus. JAMA 287:2563–2569 Writing Team for the DCCT/EDIC Research Group (2002) Effect of intensive therapy on the microvascular complications of type 1 diabetes mellitus. JAMA 287:2563–2569
20.
go back to reference EDIC study (2003) Sustained effect of intensive treatment of type 1 diabetes mellitus on development and progression of diabetic nephropathy: the Epidemiology of Diabetes Interventions and Complications (EDIC) study. JAMA 290:2159–2167 EDIC study (2003) Sustained effect of intensive treatment of type 1 diabetes mellitus on development and progression of diabetic nephropathy: the Epidemiology of Diabetes Interventions and Complications (EDIC) study. JAMA 290:2159–2167
21.
go back to reference Nathan DM, Cleary PA, Backlund JY et al (2005) Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med 353:2643–2653PubMed Nathan DM, Cleary PA, Backlund JY et al (2005) Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med 353:2643–2653PubMed
22.
go back to reference Chalmers J, Cooper ME (2008) UKPDS and the legacy effect. N Engl J Med 359:1618–1620PubMed Chalmers J, Cooper ME (2008) UKPDS and the legacy effect. N Engl J Med 359:1618–1620PubMed
23.
go back to reference Li SL, Reddy MA, Cai Q et al (2006) Enhanced proatherogenic responses in macrophages and vascular smooth muscle cells derived from diabetic db/db mice. Diabetes 55:2611–2619PubMed Li SL, Reddy MA, Cai Q et al (2006) Enhanced proatherogenic responses in macrophages and vascular smooth muscle cells derived from diabetic db/db mice. Diabetes 55:2611–2619PubMed
24.
go back to reference Villeneuve LM, Reddy MA, Lanting LL, Wang M, Meng L, Natarajan R (2008) Epigenetic histone H3 lysine 9 methylation in metabolic memory and inflammatory phenotype of vascular smooth muscle cells in diabetes. Proc Natl Acad Sci U S A 105:9047–9052PubMedCentralPubMed Villeneuve LM, Reddy MA, Lanting LL, Wang M, Meng L, Natarajan R (2008) Epigenetic histone H3 lysine 9 methylation in metabolic memory and inflammatory phenotype of vascular smooth muscle cells in diabetes. Proc Natl Acad Sci U S A 105:9047–9052PubMedCentralPubMed
25.
go back to reference Reddy MA, Jin W, Villeneuve L et al (2012) Pro-inflammatory role of microRNA-200 in vascular smooth muscle cells from diabetic mice. Arterioscler Thromb Vasc Biol 32:721–729PubMedCentralPubMed Reddy MA, Jin W, Villeneuve L et al (2012) Pro-inflammatory role of microRNA-200 in vascular smooth muscle cells from diabetic mice. Arterioscler Thromb Vasc Biol 32:721–729PubMedCentralPubMed
26.
go back to reference El-Osta A, Brasacchio D, Yao D et al (2008) Transient high glucose causes persistent epigenetic changes and altered gene expression during subsequent normoglycemia. J Exp Med 205:2409–2417PubMedCentralPubMed El-Osta A, Brasacchio D, Yao D et al (2008) Transient high glucose causes persistent epigenetic changes and altered gene expression during subsequent normoglycemia. J Exp Med 205:2409–2417PubMedCentralPubMed
27.
go back to reference Brasacchio D, Okabe J, Tikellis C et al (2009) Hyperglycaemia induces a dynamic cooperativity of histone methylase and demethylase enzymes associated with gene-activating epigenetic marks that coexist on the lysine tail. Diabetes 58:1229–1236PubMedCentralPubMed Brasacchio D, Okabe J, Tikellis C et al (2009) Hyperglycaemia induces a dynamic cooperativity of histone methylase and demethylase enzymes associated with gene-activating epigenetic marks that coexist on the lysine tail. Diabetes 58:1229–1236PubMedCentralPubMed
28.
go back to reference Kowluru RA (2003) Effect of reinstitution of good glycaemic control on retinal oxidative stress and nitrative stress in diabetic rats. Diabetes 52:818–823PubMed Kowluru RA (2003) Effect of reinstitution of good glycaemic control on retinal oxidative stress and nitrative stress in diabetic rats. Diabetes 52:818–823PubMed
29.
go back to reference Zhong Q, Kowluru RA (2013) Epigenetic modification of Sod2 in the development of diabetic retinopathy and in the metabolic memory: role of histone methylation. Invest Ophthalmol Vis Sci 54:244–250PubMedCentralPubMed Zhong Q, Kowluru RA (2013) Epigenetic modification of Sod2 in the development of diabetic retinopathy and in the metabolic memory: role of histone methylation. Invest Ophthalmol Vis Sci 54:244–250PubMedCentralPubMed
30.
go back to reference Zhong Q, Kowluru RA (2013) Regulation of matrix metalloproteinase-9 by epigenetic modifications and the development of diabetic retinopathy. Diabetes 62:2559–2568PubMedCentralPubMed Zhong Q, Kowluru RA (2013) Regulation of matrix metalloproteinase-9 by epigenetic modifications and the development of diabetic retinopathy. Diabetes 62:2559–2568PubMedCentralPubMed
31.
go back to reference Zhong Q, Kowluru RA (2011) Epigenetic changes in mitochondrial superoxide dismutase in the retina and the development of diabetic retinopathy. Diabetes 60:1304–1313PubMedCentralPubMed Zhong Q, Kowluru RA (2011) Epigenetic changes in mitochondrial superoxide dismutase in the retina and the development of diabetic retinopathy. Diabetes 60:1304–1313PubMedCentralPubMed
32.
go back to reference Engerman RL, Kern TS (1987) Progression of incipient diabetic retinopathy during good glycaemic control. Diabetes 36:808–812PubMed Engerman RL, Kern TS (1987) Progression of incipient diabetic retinopathy during good glycaemic control. Diabetes 36:808–812PubMed
33.
go back to reference Kowluru RA, Abbas SN, Odenbach S (2004) Reversal of hyperglycaemia and diabetic nephropathy: effect of reinstitution of good metabolic control on oxidative stress in the kidney of diabetic rats. J Diabetes Complications 18:282–288PubMed Kowluru RA, Abbas SN, Odenbach S (2004) Reversal of hyperglycaemia and diabetic nephropathy: effect of reinstitution of good metabolic control on oxidative stress in the kidney of diabetic rats. J Diabetes Complications 18:282–288PubMed
34.
go back to reference Kouzarides T (2007) Chromatin modifications and their function. Cell 128:693–705PubMed Kouzarides T (2007) Chromatin modifications and their function. Cell 128:693–705PubMed
35.
go back to reference Jones PA (2012) Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 13:484–492PubMed Jones PA (2012) Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet 13:484–492PubMed
36.
go back to reference Zhou VW, Goren A, Bernstein BE (2011) Charting histone modifications and the functional organization of mammalian genomes. Nat Rev Genet 12:7–18PubMed Zhou VW, Goren A, Bernstein BE (2011) Charting histone modifications and the functional organization of mammalian genomes. Nat Rev Genet 12:7–18PubMed
37.
go back to reference Portela A, Esteller M (2010) Epigenetic modifications and human disease. Nat Biotechnol 28:1057–1068PubMed Portela A, Esteller M (2010) Epigenetic modifications and human disease. Nat Biotechnol 28:1057–1068PubMed
38.
go back to reference Baylin SB, Jones PA (2011) A decade of exploring the cancer epigenome—biological and translational implications. Nat Rev Cancer 11:726–734PubMedCentralPubMed Baylin SB, Jones PA (2011) A decade of exploring the cancer epigenome—biological and translational implications. Nat Rev Cancer 11:726–734PubMedCentralPubMed
39.
go back to reference Ling C, Groop L (2009) Epigenetics: a molecular link between environmental factors and type 2 diabetes. Diabetes 58:2718–2725PubMedCentralPubMed Ling C, Groop L (2009) Epigenetics: a molecular link between environmental factors and type 2 diabetes. Diabetes 58:2718–2725PubMedCentralPubMed
40.
41.
go back to reference Sandholm N, Salem RM, McKnight AJ et al (2012) New susceptibility loci associated with kidney disease in type 1 diabetes. PLoS Genet 8:e1002921PubMedCentralPubMed Sandholm N, Salem RM, McKnight AJ et al (2012) New susceptibility loci associated with kidney disease in type 1 diabetes. PLoS Genet 8:e1002921PubMedCentralPubMed
42.
43.
go back to reference McKnight AJ, McKay GJ, Maxwell AP (2014) Genetic and epigenetic risk factors for diabetic kidney disease. Adv Chron Kidney Dis 21:287–296 McKnight AJ, McKay GJ, Maxwell AP (2014) Genetic and epigenetic risk factors for diabetic kidney disease. Adv Chron Kidney Dis 21:287–296
44.
go back to reference Maurano MT, Humbert R, Rynes E et al (2012) Systematic localization of common disease-associated variation in regulatory DNA. Science 337:1190–1195PubMedCentralPubMed Maurano MT, Humbert R, Rynes E et al (2012) Systematic localization of common disease-associated variation in regulatory DNA. Science 337:1190–1195PubMedCentralPubMed
46.
go back to reference Jirtle RL, Skinner MK (2007) Environmental epigenomics and disease susceptibility. Nat Rev Genet 8:253–262PubMed Jirtle RL, Skinner MK (2007) Environmental epigenomics and disease susceptibility. Nat Rev Genet 8:253–262PubMed
47.
go back to reference Simmons R (2011) Epigenetics and maternal nutrition: nature v. nurture. Proc Nutr Soc 70:73–81PubMed Simmons R (2011) Epigenetics and maternal nutrition: nature v. nurture. Proc Nutr Soc 70:73–81PubMed
48.
go back to reference Bell CG, Teschendorff AE, Rakyan VK, Maxwell AP, Beck S, Savage DA (2010) Genome-wide DNA methylation analysis for diabetic nephropathy in type 1 diabetes mellitus. BMC Med Gen 3:33–42 Bell CG, Teschendorff AE, Rakyan VK, Maxwell AP, Beck S, Savage DA (2010) Genome-wide DNA methylation analysis for diabetic nephropathy in type 1 diabetes mellitus. BMC Med Gen 3:33–42
49.
go back to reference Sapienza C, Lee J, Powell J et al (2011) DNA methylation profiling identifies epigenetic differences between diabetes patients with ESRD and diabetes patients without nephropathy. Epigenetics Off J DNA Methylation Soc 6:20–28 Sapienza C, Lee J, Powell J et al (2011) DNA methylation profiling identifies epigenetic differences between diabetes patients with ESRD and diabetes patients without nephropathy. Epigenetics Off J DNA Methylation Soc 6:20–28
50.
go back to reference Bechtel W, McGoohan S, Zeisberg EM et al (2010) Methylation determines fibroblast activation and fibrogenesis in the kidney. Nat Med 16:544–550PubMedCentralPubMed Bechtel W, McGoohan S, Zeisberg EM et al (2010) Methylation determines fibroblast activation and fibrogenesis in the kidney. Nat Med 16:544–550PubMedCentralPubMed
51.
go back to reference Hasegawa K, Wakino S, Simic P et al (2013) Renal tubular Sirt1 attenuates diabetic albuminuria by epigenetically suppressing Claudin-1 overexpression in podocytes. Nat Med 19:1496–1504PubMedCentralPubMed Hasegawa K, Wakino S, Simic P et al (2013) Renal tubular Sirt1 attenuates diabetic albuminuria by epigenetically suppressing Claudin-1 overexpression in podocytes. Nat Med 19:1496–1504PubMedCentralPubMed
52.
go back to reference Ko YA, Mohtat D, Suzuki M et al (2013) Cytosine methylation changes in enhancer regions of core pro-fibrotic genes characterize kidney fibrosis development. Genome Biol 14:R108PubMedCentralPubMed Ko YA, Mohtat D, Suzuki M et al (2013) Cytosine methylation changes in enhancer regions of core pro-fibrotic genes characterize kidney fibrosis development. Genome Biol 14:R108PubMedCentralPubMed
53.
go back to reference Pirola L, Balcerczyk A, Tothill RW et al (2011) Genome-wide analysis distinguishes hyperglycaemia regulated epigenetic signatures of primary vascular cells. Genome Res 21:1601–1615PubMedCentralPubMed Pirola L, Balcerczyk A, Tothill RW et al (2011) Genome-wide analysis distinguishes hyperglycaemia regulated epigenetic signatures of primary vascular cells. Genome Res 21:1601–1615PubMedCentralPubMed
54.
go back to reference Kim ES, Isoda F, Kurland I, Mobbs CV (2013) Glucose-induced metabolic memory in Schwann cells: prevention by PPAR agonists. Endocrinology 154:3054–3066PubMed Kim ES, Isoda F, Kurland I, Mobbs CV (2013) Glucose-induced metabolic memory in Schwann cells: prevention by PPAR agonists. Endocrinology 154:3054–3066PubMed
55.
56.
go back to reference Klose RJ, Zhang Y (2007) Regulation of histone methylation by demethylimination and demethylation. Nat Rev Mol Cell Biol 8:307–318PubMed Klose RJ, Zhang Y (2007) Regulation of histone methylation by demethylimination and demethylation. Nat Rev Mol Cell Biol 8:307–318PubMed
57.
go back to reference Allis CD, Berger SL, Cote J et al (2007) New nomenclature for chromatin-modifying enzymes. Cell 131:633–636PubMed Allis CD, Berger SL, Cote J et al (2007) New nomenclature for chromatin-modifying enzymes. Cell 131:633–636PubMed
58.
go back to reference Smith E, Shilatifard A (2010) The chromatin signaling pathway: diverse mechanisms of recruitment of histone-modifying enzymes and varied biological outcomes. Mol Cell 40:689–701PubMedCentralPubMed Smith E, Shilatifard A (2010) The chromatin signaling pathway: diverse mechanisms of recruitment of histone-modifying enzymes and varied biological outcomes. Mol Cell 40:689–701PubMedCentralPubMed
59.
go back to reference Rinn JL, Chang HY (2012) Genome regulation by long noncoding RNAs. Annu Rev Biochem 81:145–166PubMed Rinn JL, Chang HY (2012) Genome regulation by long noncoding RNAs. Annu Rev Biochem 81:145–166PubMed
60.
go back to reference Sassone-Corsi P (2013) Physiology. When metabolism and epigenetics converge. Science 339:148–150PubMed Sassone-Corsi P (2013) Physiology. When metabolism and epigenetics converge. Science 339:148–150PubMed
61.
go back to reference Abmayr SM, Workman JL (2012) Holding on through DNA replication: histone modification or modifier? Cell 150:875–877PubMed Abmayr SM, Workman JL (2012) Holding on through DNA replication: histone modification or modifier? Cell 150:875–877PubMed
62.
go back to reference Cedar H, Bergman Y (2009) Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet 10:295–304PubMed Cedar H, Bergman Y (2009) Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet 10:295–304PubMed
64.
go back to reference Das F, Ghosh-Choudhury N, Venkatesan B, Li X, Mahimainathan L, Choudhury GG (2008) Akt kinase targets association of CBP with SMAD 3 to regulate TGFbeta-induced expression of plasminogen activator inhibitor-1. J Cell Physiol 214:513–527PubMed Das F, Ghosh-Choudhury N, Venkatesan B, Li X, Mahimainathan L, Choudhury GG (2008) Akt kinase targets association of CBP with SMAD 3 to regulate TGFbeta-induced expression of plasminogen activator inhibitor-1. J Cell Physiol 214:513–527PubMed
65.
go back to reference Yuan H, Reddy MA, Sun G et al (2013) Involvement of p300/CBP and epigenetic histone acetylation in TGF-beta1-mediated gene transcription in mesangial cells. Am J Physiol Ren Physiol 304:F601–F613 Yuan H, Reddy MA, Sun G et al (2013) Involvement of p300/CBP and epigenetic histone acetylation in TGF-beta1-mediated gene transcription in mesangial cells. Am J Physiol Ren Physiol 304:F601–F613
66.
go back to reference Sun G, Reddy MA, Yuan H, Lanting L, Kato M, Natarajan R (2010) Epigenetic histone methylation modulates fibrotic gene expression. J Am Soc Nephrol 21:2069–2080PubMedCentralPubMed Sun G, Reddy MA, Yuan H, Lanting L, Kato M, Natarajan R (2010) Epigenetic histone methylation modulates fibrotic gene expression. J Am Soc Nephrol 21:2069–2080PubMedCentralPubMed
67.
go back to reference Komers R, Mar D, Denisenko O, Xu B, Oyama TT, Bomsztyk K (2013) Epigenetic changes in renal genes dysregulated in mouse and rat models of type 1 diabetes. Lab Investig J Tech Methods Pathol 93:543–552 Komers R, Mar D, Denisenko O, Xu B, Oyama TT, Bomsztyk K (2013) Epigenetic changes in renal genes dysregulated in mouse and rat models of type 1 diabetes. Lab Investig J Tech Methods Pathol 93:543–552
68.
go back to reference Bock F, Shahzad K, Wang H et al (2013) Activated protein C ameliorates diabetic nephropathy by epigenetically inhibiting the redox enzyme p66Shc. Proc Natl Acad Sci U S A 110:648–653PubMedCentralPubMed Bock F, Shahzad K, Wang H et al (2013) Activated protein C ameliorates diabetic nephropathy by epigenetically inhibiting the redox enzyme p66Shc. Proc Natl Acad Sci U S A 110:648–653PubMedCentralPubMed
69.
go back to reference Chen J, Guo Y, Zeng W et al (2014) ER stress triggers MCP-1 expression through SET7/9-induced histone methylation in the kidneys of db/db mice. Am J Physiol Ren Physiol 306:F916–F925 Chen J, Guo Y, Zeng W et al (2014) ER stress triggers MCP-1 expression through SET7/9-induced histone methylation in the kidneys of db/db mice. Am J Physiol Ren Physiol 306:F916–F925
70.
go back to reference Reddy MA, Sumanth P, Lanting L et al (2014) Losartan reverses permissive epigenetic changes in renal glomeruli of diabetic db/db mice. Kidney Int 85:362–373PubMedCentralPubMed Reddy MA, Sumanth P, Lanting L et al (2014) Losartan reverses permissive epigenetic changes in renal glomeruli of diabetic db/db mice. Kidney Int 85:362–373PubMedCentralPubMed
71.
go back to reference Ruggenenti P, Cravedi P, Remuzzi G (2010) The RAAS in the pathogenesis and treatment of diabetic nephropathy. Nat Rev Nephrol 6:319–330PubMed Ruggenenti P, Cravedi P, Remuzzi G (2010) The RAAS in the pathogenesis and treatment of diabetic nephropathy. Nat Rev Nephrol 6:319–330PubMed
72.
go back to reference Perrone L, Devi TS, Hosoya K, Terasaki T, Singh LP (2009) Thioredoxin interacting protein (TXNIP) induces inflammation through chromatin modification in retinal capillary endothelial cells under diabetic conditions. J Cell Physiol 221:262–272PubMed Perrone L, Devi TS, Hosoya K, Terasaki T, Singh LP (2009) Thioredoxin interacting protein (TXNIP) induces inflammation through chromatin modification in retinal capillary endothelial cells under diabetic conditions. J Cell Physiol 221:262–272PubMed
73.
go back to reference Kadiyala CS, Zheng L, Du Y et al (2012) Acetylation of retinal histones in diabetes increases inflammatory proteins: effects of minocycline and manipulation of histone acetyltransferase (HAT) and histone deacetylase (HDAC). J Biol Chem 287:25869–25880PubMedCentralPubMed Kadiyala CS, Zheng L, Du Y et al (2012) Acetylation of retinal histones in diabetes increases inflammatory proteins: effects of minocycline and manipulation of histone acetyltransferase (HAT) and histone deacetylase (HDAC). J Biol Chem 287:25869–25880PubMedCentralPubMed
74.
go back to reference Kaur H, Chen S, Xin X, Chiu J, Khan ZA, Chakrabarti S (2006) Diabetes-induced extracellular matrix protein expression is mediated by transcription coactivator p300. Diabetes 55:3104–3111PubMed Kaur H, Chen S, Xin X, Chiu J, Khan ZA, Chakrabarti S (2006) Diabetes-induced extracellular matrix protein expression is mediated by transcription coactivator p300. Diabetes 55:3104–3111PubMed
75.
go back to reference Feng B, Chen S, Chiu J, George B, Chakrabarti S (2008) Regulation of cardiomyocyte hypertrophy in diabetes at the transcriptional level. Am J Physiol Endocrinol Metab 294:E1119–E1126PubMed Feng B, Chen S, Chiu J, George B, Chakrabarti S (2008) Regulation of cardiomyocyte hypertrophy in diabetes at the transcriptional level. Am J Physiol Endocrinol Metab 294:E1119–E1126PubMed
76.
go back to reference Rafehi H, El-Osta A, Karagiannis TC (2011) Genetic and epigenetic events in diabetic wound healing. Int Wound J 8:12–21PubMed Rafehi H, El-Osta A, Karagiannis TC (2011) Genetic and epigenetic events in diabetic wound healing. Int Wound J 8:12–21PubMed
77.
go back to reference Miao F, Gonzalo IG, Lanting L, Natarajan R (2004) In vivo chromatin remodeling events leading to inflammatory gene transcription under diabetic conditions. J Biol Chem 279:18091–18097PubMed Miao F, Gonzalo IG, Lanting L, Natarajan R (2004) In vivo chromatin remodeling events leading to inflammatory gene transcription under diabetic conditions. J Biol Chem 279:18091–18097PubMed
78.
go back to reference Miao F, Wu X, Zhang L, Yuan YC, Riggs AD, Natarajan R (2007) Genome-wide analysis of histone lysine methylation variations caused by diabetic conditions in human monocytes. J Biol Chem 282:13854–13863PubMed Miao F, Wu X, Zhang L, Yuan YC, Riggs AD, Natarajan R (2007) Genome-wide analysis of histone lysine methylation variations caused by diabetic conditions in human monocytes. J Biol Chem 282:13854–13863PubMed
79.
go back to reference Miao F, Smith DD, Zhang L, Min A, Feng W, Natarajan R (2008) Lymphocytes from patients with type 1 diabetes display a distinct profile of chromatin histone H3 lysine 9 dimethylation: an epigenetic study in diabetes. Diabetes 57:3189–3198PubMedCentralPubMed Miao F, Smith DD, Zhang L, Min A, Feng W, Natarajan R (2008) Lymphocytes from patients with type 1 diabetes display a distinct profile of chromatin histone H3 lysine 9 dimethylation: an epigenetic study in diabetes. Diabetes 57:3189–3198PubMedCentralPubMed
80.
go back to reference Miao F, Chen Z, Zhang L et al (2012) Profiles of epigenetic histone post-translational modifications at type 1 diabetes susceptible genes. J Biol Chem 287:16335–16345PubMedCentralPubMed Miao F, Chen Z, Zhang L et al (2012) Profiles of epigenetic histone post-translational modifications at type 1 diabetes susceptible genes. J Biol Chem 287:16335–16345PubMedCentralPubMed
81.
go back to reference Li Y, Reddy MA, Miao F et al (2008) Role of the histone H3 lysine 4 methyltransferase, SET7/9, in the regulation of NF-κB-dependent inflammatory genes. Relevance to diabetes and inflammation. J Biol Chem 283:26771–26781PubMedCentralPubMed Li Y, Reddy MA, Miao F et al (2008) Role of the histone H3 lysine 4 methyltransferase, SET7/9, in the regulation of NF-κB-dependent inflammatory genes. Relevance to diabetes and inflammation. J Biol Chem 283:26771–26781PubMedCentralPubMed
82.
go back to reference Okabe J, Orlowski C, Balcerczyk A et al (2012) Distinguishing hyperglycaemic changes by Set7 in vascular endothelial cells. Circ Res 110:1067–1076PubMed Okabe J, Orlowski C, Balcerczyk A et al (2012) Distinguishing hyperglycaemic changes by Set7 in vascular endothelial cells. Circ Res 110:1067–1076PubMed
83.
go back to reference Okabe J, Fernandez AZ, Ziemann M, Keating ST, Balcerczyk A, El-Osta A (2014) Endothelial transcriptome in response to pharmacological methyltransferase inhibition. ChemMedChem 9:1755–1762PubMed Okabe J, Fernandez AZ, Ziemann M, Keating ST, Balcerczyk A, El-Osta A (2014) Endothelial transcriptome in response to pharmacological methyltransferase inhibition. ChemMedChem 9:1755–1762PubMed
84.
go back to reference Villeneuve LM, Kato M, Reddy MA, Wang M, Lanting L, Natarajan R (2010) Enhanced levels of microRNA-125b in vascular smooth muscle cells of diabetic db/db mice lead to increased inflammatory gene expression by targeting the histone methyltransferase Suv39h1. Diabetes 59:2904–2915PubMedCentralPubMed Villeneuve LM, Kato M, Reddy MA, Wang M, Lanting L, Natarajan R (2010) Enhanced levels of microRNA-125b in vascular smooth muscle cells of diabetic db/db mice lead to increased inflammatory gene expression by targeting the histone methyltransferase Suv39h1. Diabetes 59:2904–2915PubMedCentralPubMed
85.
go back to reference Miao F, Chen Z, Genuth S et al (2014) Evaluating the role of epigenetic histone modifications in the metabolic memory of type 1 diabetes. Diabetes 63:1748–1762PubMed Miao F, Chen Z, Genuth S et al (2014) Evaluating the role of epigenetic histone modifications in the metabolic memory of type 1 diabetes. Diabetes 63:1748–1762PubMed
86.
go back to reference Guttman M, Amit I, Garber M et al (2009) Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 458:223–227PubMedCentralPubMed Guttman M, Amit I, Garber M et al (2009) Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 458:223–227PubMedCentralPubMed
87.
go back to reference Alvarez ML, Distefano JK (2013) The role of non-coding RNAs in diabetic nephropathy: potential applications as biomarkers for disease development and progression. Diabetes Res Clin Pract 99:1–11PubMed Alvarez ML, Distefano JK (2013) The role of non-coding RNAs in diabetic nephropathy: potential applications as biomarkers for disease development and progression. Diabetes Res Clin Pract 99:1–11PubMed
88.
go back to reference Kato M, Castro NE, Natarajan R (2013) MicroRNAs: potential mediators and biomarkers of diabetic complications. Free Radic Biol Med 64:85–94PubMedCentralPubMed Kato M, Castro NE, Natarajan R (2013) MicroRNAs: potential mediators and biomarkers of diabetic complications. Free Radic Biol Med 64:85–94PubMedCentralPubMed
89.
go back to reference Kantharidis P, Wang B, Carew RM, Lan HY (2011) Diabetes complications: the microRNA perspective. Diabetes 60:1832–1837PubMedCentralPubMed Kantharidis P, Wang B, Carew RM, Lan HY (2011) Diabetes complications: the microRNA perspective. Diabetes 60:1832–1837PubMedCentralPubMed
91.
92.
go back to reference Cabili MN, Trapnell C, Goff L et al (2011) Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 25:1915–1927PubMedCentralPubMed Cabili MN, Trapnell C, Goff L et al (2011) Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev 25:1915–1927PubMedCentralPubMed
93.
go back to reference Kato M, Putta S, Wang M et al (2009) TGF-β activates Akt kinase through a microRNA-dependent amplifying circuit targeting PTEN. Nat Cell Biol 11:881–889PubMedCentralPubMed Kato M, Putta S, Wang M et al (2009) TGF-β activates Akt kinase through a microRNA-dependent amplifying circuit targeting PTEN. Nat Cell Biol 11:881–889PubMedCentralPubMed
94.
go back to reference Alvarez ML, DiStefano JK (2011) Functional characterization of the plasmacytoma variant translocation 1 gene (PVT1) in diabetic nephropathy. PLoS One 6:e18671PubMedCentralPubMed Alvarez ML, DiStefano JK (2011) Functional characterization of the plasmacytoma variant translocation 1 gene (PVT1) in diabetic nephropathy. PLoS One 6:e18671PubMedCentralPubMed
95.
go back to reference Hanson RL, Craig DW, Millis MP et al (2007) Identification of PVT1 as a candidate gene for end-stage renal disease in type 2 diabetes using a pooling-based genome-wide single nucleotide polymorphism association study. Diabetes 56:975–983PubMed Hanson RL, Craig DW, Millis MP et al (2007) Identification of PVT1 as a candidate gene for end-stage renal disease in type 2 diabetes using a pooling-based genome-wide single nucleotide polymorphism association study. Diabetes 56:975–983PubMed
96.
go back to reference Leung A, Trac C, Jin W et al (2013) Novel long noncoding RNAs Are regulated by angiotensin II in vascular smooth muscle cells. Circ Res 113:266–278PubMedCentralPubMed Leung A, Trac C, Jin W et al (2013) Novel long noncoding RNAs Are regulated by angiotensin II in vascular smooth muscle cells. Circ Res 113:266–278PubMedCentralPubMed
97.
go back to reference Reddy MA, Chen Z, Park JT et al (2014) Regulation of inflammatory phenotype in macrophages by a diabetes-induced long non-coding RNA. Diabetes. doi:10.2337/db14-0298 Reddy MA, Chen Z, Park JT et al (2014) Regulation of inflammatory phenotype in macrophages by a diabetes-induced long non-coding RNA. Diabetes. doi:10.​2337/​db14-0298
98.
go back to reference Mali P, Aach J, Stranges PB et al (2013) CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat Biotechnol 31:833–838PubMed Mali P, Aach J, Stranges PB et al (2013) CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat Biotechnol 31:833–838PubMed
99.
100.
go back to reference Laird PW (2010) Principles and challenges of genome-wide DNA methylation analysis. Nat Rev Genet 11:191–203PubMed Laird PW (2010) Principles and challenges of genome-wide DNA methylation analysis. Nat Rev Genet 11:191–203PubMed
101.
go back to reference Simon JM, Giresi PG, Davis IJ, Lieb JD (2012) Using formaldehyde-assisted isolation of regulatory elements (FAIRE) to isolate active regulatory DNA. Nat Protoc 7:256–267PubMedCentralPubMed Simon JM, Giresi PG, Davis IJ, Lieb JD (2012) Using formaldehyde-assisted isolation of regulatory elements (FAIRE) to isolate active regulatory DNA. Nat Protoc 7:256–267PubMedCentralPubMed
102.
go back to reference Leung A, Parks BW, Du J et al (2014) Open chromatin profiling in mice livers reveals unique chromatin variations induced by high fat diet. J Biol Chem 289:23557–23567PubMed Leung A, Parks BW, Du J et al (2014) Open chromatin profiling in mice livers reveals unique chromatin variations induced by high fat diet. J Biol Chem 289:23557–23567PubMed
103.
go back to reference Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B (2008) Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods 5:621–628PubMed Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B (2008) Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods 5:621–628PubMed
104.
go back to reference Mouse ENCODE Consortium, Stamatoyannopoulos JA, Snyder M et al (2012) An encyclopedia of mouse DNA elements (Mouse ENCODE). Genome Biol 13:418PubMedCentralPubMed Mouse ENCODE Consortium, Stamatoyannopoulos JA, Snyder M et al (2012) An encyclopedia of mouse DNA elements (Mouse ENCODE). Genome Biol 13:418PubMedCentralPubMed
105.
go back to reference Hodgin JB, Nair V, Zhang H et al (2013) Identification of cross-species shared transcriptional networks of diabetic nephropathy in human and mouse glomeruli. Diabetes 62:299–308PubMedCentralPubMed Hodgin JB, Nair V, Zhang H et al (2013) Identification of cross-species shared transcriptional networks of diabetic nephropathy in human and mouse glomeruli. Diabetes 62:299–308PubMedCentralPubMed
106.
go back to reference Ronn T, Ling C (2013) Effect of exercise on DNA methylation and metabolism in human adipose tissue and skeletal muscle. Epigenomics 5:603–605PubMed Ronn T, Ling C (2013) Effect of exercise on DNA methylation and metabolism in human adipose tissue and skeletal muscle. Epigenomics 5:603–605PubMed
Metadata
Title
Epigenetic mechanisms in diabetic complications and metabolic memory
Authors
Marpadga A. Reddy
Erli Zhang
Rama Natarajan
Publication date
01-03-2015
Publisher
Springer Berlin Heidelberg
Published in
Diabetologia / Issue 3/2015
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-014-3462-y

Other articles of this Issue 3/2015

Diabetologia 3/2015 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.